1
|
Tao MH, Chen S, Freudenheim JL, Cauley JA, Johnson KC, Mai X, Sarto GE, Wakelee H, Boffetta P, Wactawski-Wende J. Oral bisphosphonate use and lung cancer incidence among postmenopausal women. Ann Oncol 2019; 29:1476-1485. [PMID: 29617712 PMCID: PMC6005043 DOI: 10.1093/annonc/mdy097] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background Bisphosphonates are common medications for the treatment of osteoporosis in older populations. Several studies, including the Women’s Health Initiative (WHI), have found inverse associations of bisphosphonate use with risk of breast and endometrial cancer, but little is known about its association with other common malignancies. The objective of this study was to evaluate the association of bisphosphonate use on the incidence of lung cancer in the WHI. Patients and methods The association between oral bisphosphonate use and lung cancer risk was examined in 151 432 postmenopausal women enrolled into the WHI in 1993–1998. At baseline and during follow-up, participants completed an inventory of regularly used medications including bisphosphonates. Results After a mean follow-up of 13.3 years, 2511 women were diagnosed with incident lung cancer. There was no evidence of a difference in lung cancer incidence between oral bisphosphonate users and never users (adjusted hazard ratio = 0.91; 95% confidence intervals, 0.80–1.04; P = 0.16). However, an inverse association was observed among those who were never smokers (hazard ratio = 0.57, 95% confidence interval, 0.39–0.84; P < 0.01). Conclusion In this large prospective cohort of postmenopausal women, oral bisphosphonate use was associated with significantly lower lung cancer risk among never smokers, suggesting bisphosphonates may have a protective effect against lung cancer. Additional studies are needed to confirm our findings.
Collapse
Affiliation(s)
- M H Tao
- Department of Biostatistics and Epidemiology, University of North Texas Health Science Center, Fort Worth, USA.
| | - S Chen
- Department of Biostatistics and Epidemiology, University of North Texas Health Science Center, Fort Worth, USA
| | - J L Freudenheim
- Department of Epidemiology and Environmental Health, University at Buffalo, Buffalo, USA
| | - J A Cauley
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, USA
| | - K C Johnson
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, USA
| | - X Mai
- Department of Epidemiology and Environmental Health, University at Buffalo, Buffalo, USA
| | - G E Sarto
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, USA
| | - H Wakelee
- Division of Oncology, Department of Medicine, Stanford University and Stanford Cancer Institute, Stanford, USA
| | - P Boffetta
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - J Wactawski-Wende
- Department of Epidemiology and Environmental Health, University at Buffalo, Buffalo, USA
| |
Collapse
|
2
|
Park YE, Bava U, Lin JM, Cornish J, Naot D, Reid IR. Bone-Bound Bisphosphonates Inhibit Proliferation of Breast Cancer Cells. Calcif Tissue Int 2019; 105:497-505. [PMID: 31324954 DOI: 10.1007/s00223-019-00590-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/12/2019] [Indexed: 12/31/2022]
Abstract
Bisphosphonates are used in treating patients with breast cancer. In vitro studies have shown that bisphosphonates act directly on tumour cells, inhibiting cell proliferation and inducing apoptosis. In most such studies, drugs were added to culture media exposing cells to high bisphosphonate concentrations in solution. However, since bisphosphonates bind to bone hydroxyapatite with high affinity and remain bound for very long periods of time, these experimental systems are not an optimal model for the action of the drugs in vivo. The aim of this study was to determine whether bone-bound zoledronate has direct effects on adjacent breast cancer cells. Bone slices were pre-incubated with bisphosphonate solutions, washed, and seeded with cells of the breast cancer cell lines, MCF7 or MDA-MB-231. Proliferation was assessed by cell counts and thymidine incorporation for up to 72 h. Inhibition of the mevalonate pathway was tested by measuring the levels of unprenylated Rap1A, and apoptosis was examined by the presence of cleaved caspase-8 on western blots. The proliferation rate of breast cancer cells on zoledronate-treated bone was significantly lower compared to cells on control bone. Other bisphosphonates showed a similar inhibitory effect, with an order of potency similar to their clinical potencies. Unprenylated Rap1A accumulated in MCF7 cells on zoledronate-treated bone, suggesting zoledronate acted through the inhibition of the mevalonate pathway. Accumulation of cleaved caspase-8 in MDA-MB-231 cells on bisphosphonate-treated bone indicated increased apoptosis in the cells. In conclusion, bone-bound zoledronate inhibits breast cancer cell proliferation, an activity that may contribute to its clinical anti-tumour effects.
Collapse
Affiliation(s)
- Young-Eun Park
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Usha Bava
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Jian-Ming Lin
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Jillian Cornish
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Dorit Naot
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Ian R Reid
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| |
Collapse
|
3
|
Cui X, Li S, Gu J, Lin Z, Lai B, Huang L, Feng J, Liu B, Zhou Y. Retrospective study on the efficacy of bisphosphonates in tyrosine kinase inhibitor-treated patients with non-small cell lung cancer exhibiting bone metastasis. Oncol Lett 2019; 18:5437-5447. [PMID: 31612052 PMCID: PMC6781563 DOI: 10.3892/ol.2019.10870] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 08/14/2019] [Indexed: 01/04/2023] Open
Abstract
Bisphosphonates (Bps) inhibit the maturation of osteoclasts and suppress the adhesion of cancer cells to the bone matrix. They are recommended as the standard treatment for tumors exhibiting bone metastasis (BM). However, whether Bps can improve the prognosis of patients with tyrosine kinase inhibitor (TKI)-treated non-small cell lung cancer (NSCLC) exhibiting BM remains unclear. A total of 129 patients with NSCLC initially diagnosed with BM at The First Affiliated Hospital of Sun Yat-Sen University (Guangzhou, China) between January 2005 and December 2017 were analyzed in the present retrospective study. Median progression free survival (mPFS) time, median bone metastasis overall survival (mBM-OS) time and bone-associated progression-free survival were analyzed. Among the 129 patients, patients treated with Bps experienced significantly prolonged PFS time [mPFS: 7.1 vs. 5.1 months; hazard ratio (HR), 0.51; confidence interval (CI), 0.30-0.87; P=0.0114] in comparison with patients not treated with Bps. Of the 49 patients treated with frontline TKIs (EGFR TKIs or ALK TKI), 32 received Bps at the same time, while 17 patients received TKIs alone. The results revealed that mPFS time was significantly greater in the TKIs plus Bps group than in the TKIs alone group (mPFS: 11.2 vs. 6.9 months; HR, 0.13; CI, 0.05-0.35; P<0.0001). Significantly prolonged BM-OS time was also observed in the combination group in comparison with the TKIs alone group (mBM-OS: 31 vs. 22 months; HR, 0.31; CI, 0.10-0.96; P=0.0413). The present study demonstrated that among the patients who received TKIs (EGFR TKIs or ALK TKIs), those who also received Bps experienced significantly longer PFS time and tended to exhibit significantly improved BM-OS time, which indicated that Bps should be added to the treatment regimen of patients with NSCLC exhibiting genetic mutations and bone metastasis who have been prescribed TKIs (EGFR TKIs or ALK TKIs).
Collapse
Affiliation(s)
- Xiaoxian Cui
- Department of Respiratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Shaoli Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jincui Gu
- Department of Respiratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Ziying Lin
- Department of Respiratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Bipeng Lai
- Department of Respiratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Lixia Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jinlun Feng
- Department of Respiratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Baomo Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yanbin Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
4
|
You Y, Wang Q, Li H, Ma Y, Deng Y, Ye Z, Bai F. Zoledronic acid exhibits radio-sensitizing activity in human pancreatic cancer cells via inactivation of STAT3/NF-κB signaling. Onco Targets Ther 2019; 12:4323-4330. [PMID: 31239706 PMCID: PMC6556542 DOI: 10.2147/ott.s202516] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/09/2019] [Indexed: 01/28/2023] Open
Abstract
Background: Although pancreatic cancer is typically radio-sensitive, local treatment failure and metastasis are commonly caused by the development of resistance to radiotherapy. In the current study, the radio-sensitizing actions of zoledronic acid (ZOL) on pancreatic cancer cells were investigated. Materials and methods: Three human pancreatic cancer cell lines were exposed to ZOL, ionizing radiation (IR), or a combination of both, and the effects of the respective drug regimens on cell proliferation and invasion were examined. Results: Combined treatment with low doses of ZOL plus IR efficiently increased cell death and attenuated cell invasion compared with the individual use of ZOL or IR. These effects of ZOL were associated with inactivation of signal transducer and activator of transcription 3 (STAT3) and nuclear factor-κB (NF-κB). Conclusion: Collectively, these data suggest that ZOL in combination with IR is a promising therapeutic strategy for enhancing radio-sensitivity in pancreatic cancer cells via downregulation of the STAT3/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yanjie You
- Department of Gastroenterology, Ningxia Hui Autonomous Region People's Hospital, Yinchuan, Ningxia Hui Autonomous Region 750021, People's Republic of China
| | - Qiang Wang
- Department of Science and Education, Ningxia Hui Autonomous Region People's Hospital, Yinchuan 750021, People's Republic of China
| | - Haijun Li
- Department of Radiation Oncology, The Second People's Hospital of Neijiang, Neijiang, Sichuan 641003, People's Republic of China
| | - Yuhong Ma
- Department of Gastroenterology, Ningxia Hui Autonomous Region People's Hospital, Yinchuan, Ningxia Hui Autonomous Region 750021, People's Republic of China
| | - Yanhong Deng
- Department of Gastroenterology, Ningxia Hui Autonomous Region People's Hospital, Yinchuan, Ningxia Hui Autonomous Region 750021, People's Republic of China
| | - Zhengcai Ye
- Endoscopy Center, Ningxia Hui Autonomous Region People's Hospital, Yinchuan 750021, People's Republic of China
| | - Feihu Bai
- Department of Gastroenterology, Ningxia Hui Autonomous Region People's Hospital, Yinchuan, Ningxia Hui Autonomous Region 750021, People's Republic of China
| |
Collapse
|
5
|
de Fonseka D, Morley A, Stadon L, Keenan E, Walker S, Smith S, Harvey JE, Cox RA, Dangoor A, Comins C, Rogers C, Edey A, Addeo A, Maskell NA. Zoledronic acid in the management of mesothelioma - a feasibility study (Zol-A Trial): study protocol for a randomised controlled trial. Trials 2018; 19:467. [PMID: 30157910 PMCID: PMC6116562 DOI: 10.1186/s13063-018-2851-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 08/09/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Nitrogen containing bisphosphonates such as zoledronic acid (ZA) are known to contain certain anti-cancer properties. These have been investigated in the past in various cancers such as breast, prostate and colon. ZA in particular has shown promising results in pre-clinical studies. We propose a multicentre double-blind randomised controlled feasibility study to assess the recruitment and acceptability of ZA/placebo alongside chemotherapy in malignant pleural mesothelioma (MPM). METHODS Patients will be recruited for a 13-month period from October 2016 to November 2017. Eligible patients will be identified via the regional mesothelioma multidisciplinary team meeting. Those who receive chemotherapy will be randomised to receive either ZA or placebo alongside their chemotherapy. Those who decline chemotherapy will be offered to join the trial on the non-randomised open-labelled arm of the trial. Patients will receive a maximum of six cycles of ZA/placebo, at three-weekly cycles. All patients will be followed up for six months from randomisation. Semi-structured interviews to gather data on acceptability of trial procedures, tolerability of ZA and other relevant information will take place after the participants have completed their six cycles of treatment. For a better understanding about non-participation in mesothelioma trials we also aim to interview those who decline to take part in the trial. DISCUSSION The qualitative and quantitative data gathered in this feasibility trial will hopefully pave the way to designing a robust full phase III trial to investigate the potential synergistic effect of ZA and current standard treatment for MPM, cisplatin-pemetrexed combination chemotherapy. TRIAL REGISTRATION ISRCTN Registry, ISRCTN45536692 . Registered on 9 August 2016. EudraCT no. 2015-004433-26.
Collapse
Affiliation(s)
| | - Anna Morley
- Respiratory Research Unit, North Bristol NHS Trust, Bristol, UK
| | - Louise Stadon
- Respiratory Research Unit, North Bristol NHS Trust, Bristol, UK
| | - Emma Keenan
- Respiratory Research Unit, North Bristol NHS Trust, Bristol, UK
| | - Steven Walker
- Academic Respiratory Unit, University of Bristol, Bristol, UK
| | | | - John E. Harvey
- Academic Respiratory Unit, University of Bristol, Bristol, UK
| | - R. Ashley Cox
- Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | - Adam Dangoor
- Bristol Haematology and Oncology Centre, Bristol Royal Infirmary, Bristol, UK
| | - Charles Comins
- Bristol Haematology and Oncology Centre, Bristol Royal Infirmary, Bristol, UK
| | | | - Anthony Edey
- Radiology Department, North Bristol NHS Trust, Bristol, UK
| | - Alfredo Addeo
- Bristol Haematology and Oncology Centre, Bristol Royal Infirmary, Bristol, UK
| | - Nick A. Maskell
- Academic Respiratory Unit, University of Bristol, Bristol, UK
| |
Collapse
|
6
|
Kenessey I, Kói K, Horváth O, Cserepes M, Molnár D, Izsák V, Dobos J, Hegedűs B, Tóvári J, Tímár J. KRAS-mutation status dependent effect of zoledronic acid in human non-small cell cancer preclinical models. Oncotarget 2018; 7:79503-79514. [PMID: 27780929 PMCID: PMC5346731 DOI: 10.18632/oncotarget.12806] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 09/27/2016] [Indexed: 11/25/2022] Open
Abstract
Background In non-small cell lung cancer (NSCLC) KRAS-mutant status is a negative prognostic and predictive factor. Nitrogen-containing bisphosphonates inhibit prenylation of small G-proteins (e.g. Ras, Rac, Rho) and thus may affect proliferation and migration. In our preclinical work, we investigated the effect of an aminobisphosphonate compound (zoledronic acid) on mutant and wild type KRAS-expressing human NSCLC cell lines. Results We confirmed that zoledronic acid was unable to inhibit the prenylation of mutant K-Ras unlike in the case of wild type K-Ras. In case of in vitro proliferation, the KRAS-mutant human NSCLC cell lines showed resistance to zoledronic acid wild-type KRAS-cells proved to be sensitive. Combinatory application of zoledronic acid enhanced the cytostatic effect of cisplatin. Zoledronic acid did not induce significant apoptosis. In xenograft model, zoledronic acid significantly reduced the weight of wild type KRAS-EGFR-expressing xenograft tumor by decreasing the proliferative capacity. Futhermore, zoledronic acid induced VEGF expression and improved in vivo tumor vascularization. Materials and methods Membrane association of K-Ras was examined by Western-blot. In vitro cell viability, apoptotic cell death and migration were measured in NSCLC lines with different molecular background. The in vivo effect of zoledronic acid was investigated in a SCID mouse subcutaneous xenograft model. Conclusions The in vitro and in vivo inhibitory effect of zoledronic acid was based on the blockade of cell cycle in wild type KRAS-expressing human NSCLC cells. The zoledronic acid induced vascularization supported in vivo cytostatic effect. Our preclinical investigation suggests that patients with wild type KRAS-expressing NSCLC could potentially benefit from aminobisphosphonate therapy.
Collapse
Affiliation(s)
- István Kenessey
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary.,National Cancer Registry, National Institute of Oncology, Budapest, Hungary
| | - Krisztina Kói
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Orsolya Horváth
- Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary
| | - Mihály Cserepes
- Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary.,Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Dávid Molnár
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Vera Izsák
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | | | - Balázs Hegedűs
- Hungarian Academy of Sciences-Semmelweis University Molecular Oncology Research Group, Budapest, Hungary
| | - József Tóvári
- Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary
| | - József Tímár
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary.,Hungarian Academy of Sciences-Semmelweis University Molecular Oncology Research Group, Budapest, Hungary
| |
Collapse
|
7
|
Lei W, Zeng D, Liu G, Zhu Y, Wang J, Wu H, Jiang J, Huang J. Crucial role of OX40/OX40L signaling in a murine model of asthma. Mol Med Rep 2018; 17:4213-4220. [PMID: 29344664 PMCID: PMC5802192 DOI: 10.3892/mmr.2018.8453] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 08/31/2017] [Indexed: 12/15/2022] Open
Abstract
The aim of the present study was to explore the roles of OX40/OX40 ligand (OX40L) signaling and OX40+ T cells in ovalbumin (OVA)-induced mouse asthma model. Asthma was induced by OVA exposure and subsequent co-treatment with OX40L protein, neutralizing anti-OX40L blocking antibody, OX40+ T cells or PBS. The protein expression levels of interleukin (IL)-4, IL-6, IL-13, IL-17, tumor necrosis factor (TNF)-α and interferon (IFN)-γ in bronchoalveolar lavage fluid (BALF) were examined using murine cytokine-specific ELISA. Eosinophil accumulation as well as proliferation and apoptosis of T cells in BALF were detected by Cell Counting kit-8 and flow cytometric assays. Expression of the apoptosis-related protein cleaved caspase-3 was examined in OX40+ T cells using western blot assay. Flow cytometric analysis revealed that OVA-treated mice that were co-treated with OX40L or OX40+ T cells exhibited higher eosinophil infiltration compared with control mice treated only with OVA, whereas neutralizing anti-OX40L blocking antibody inhibited eosinophil infiltration. ELISA assays demonstrated that the expression of IL-4, IL-6, IL-13, IL-17, TNF-α and IFN-γ in BALF in OX40L-treated and OX40+ T cell-treated mice was increased compared with expression levels in control mice. Treatment with OX40L protein effectively reduced apoptosis of T cells and the expression of cleaved caspase-3 in T cells. OX40L-treated and OX40+ T cell-treated mice exhibited increased asthma through OX40/OX40L signaling, which probably promoted inflammatory factor expression, eosinophil infiltration and T cell proliferation.
Collapse
Affiliation(s)
- Wei Lei
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Daxiong Zeng
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Gaoqin Liu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yehan Zhu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jiajia Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Hongya Wu
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Junhong Jiang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jianan Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
8
|
Li X, Naguib YW, Cui Z. In vivo distribution of zoledronic acid in a bisphosphonate-metal complex-based nanoparticle formulation synthesized by a reverse microemulsion method. Int J Pharm 2017; 526:69-76. [PMID: 28455136 DOI: 10.1016/j.ijpharm.2017.04.053] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/20/2017] [Accepted: 04/22/2017] [Indexed: 12/11/2022]
Abstract
Bisphosphonates are used to treat bone diseases such as osteoporosis and cancer-induced bone pain and fractures. It is thought that modifying the pharmacokinetics and biodistribution profiles of bisphosphonates (i.e. rapid renal clearance and extensive bone absorption) will not only reduce their side effects, but also expand their clinical applications to extraskeletal tissues. In the present work, using zoledronic acid (Zol) and calcium as model bisphosphonate and metal molecules, respectively, we prepared DOPA (an anionic lipid)-coated spherical Zol-Ca nanocomposites (Zol-Ca@DOPA) and developed Zol-nanoparticle formulations (i.e. Zol-Ca@bi-lipid NPs) based on the nanocomposites. The influence of the inputted weight ratio of Zol-Ca@DOPA to DSPE-PEG2k on the properties (e.g. size, size distribution, loading efficiency, encapsulation efficiency, zeta potential, and polydispersity) of Zol-Ca@bi-lipid NPs was investigated, and a type of Zol-Ca@bi-lipid NPs with size around 25nm was selected for further studies. In a mouse model, the Zol-Ca@bi-lipid NPs significantly reduced the bone distribution of Zol, increased the blood circulating time of Zol, and altered the distribution of Zol in major organs, as compared to free Zol. It is expected that similar nanoparticles prepared with bisphosphonate-metal complexes can be explored to expand the applications to bisphosphonates in extraskeletal tissues.
Collapse
Affiliation(s)
- Xu Li
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Youssef W Naguib
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Zhengrong Cui
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States; Inner Mongolia Medical University, Inner Mongolia Key Laboratory of Molecular Biology, Hohhot, Inner Mongolia, China.
| |
Collapse
|
9
|
Zhang G, Cheng R, Zhang Z, Jiang T, Ren S, Ma Z, Zhao S, Zhou C, Zhang J. Bisphosphonates enhance antitumor effect of EGFR-TKIs in patients with advanced EGFR mutant NSCLC and bone metastases. Sci Rep 2017; 7:42979. [PMID: 28211502 PMCID: PMC5314405 DOI: 10.1038/srep42979] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/17/2017] [Indexed: 12/13/2022] Open
Abstract
Whether bisphosphonates could enhance the effect of epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) in non-small-cell lung cancer (NSCLC) patients with EGFR mutation and bone metastases (BM) remains unknown. EGFR mutation status were collected from 1560 patients with NSCLC and BM. 356 NSCLC patients with EGFR mutation and BM were identified. Among them, 91 patients received EGFR-TKIs alone and 105 patients received EGFR-TKIs plus bisphosphonates as first-line therapy. Comparing to TKIs alone, EGFR-TKIs plus bisphosphonates had a statistically significant longer progression-free survival (PFS: 11.6 vs. 9.3 months; HR = 0.68, P = 0.009), while a similar overall survival (OS: 20.5 vs. 19.5 months; HR = 0.95, P = 0.743) in patients with EGFR-mutant NSCLC and BM. The incidence of skeletal-related events in combined group was numerically lower than that in EGFR-TKIs alone group (29.7% vs. 39.4%, P = 0.147). In multivariate analysis, EGFR mutation was found to be a significant independent prognostic factor for OS in NSCLC patients with BM (HR = 0.710, P = 0.021). In conclusion, EGFR mutation was the significant independent prognostic factor for OS and the addition of bisphosphonates to EGFR-TKIs could enhance the antitumor effect of EGFR-TKIs in patients with EGFR-mutant NSCLC and BM.
Collapse
Affiliation(s)
- Guowei Zhang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450003, China
| | - Ruirui Cheng
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200433, China
| | - Zengli Zhang
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200433, China
- Department of Respiratory, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200433, China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200433, China
| | - Zhiyong Ma
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450003, China
| | - Sha Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200433, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jun Zhang
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
10
|
Li YY, Wu C, Chen SM, Shah SS, Wangpaichitr M, Feun LG, Kuo MT, Suarez M, Prince J, Savaraj N. BRAF inhibitor resistance enhances vulnerability to arginine deprivation in melanoma. Oncotarget 2017; 7:17665-80. [PMID: 26771234 PMCID: PMC4951241 DOI: 10.18632/oncotarget.6882] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/03/2016] [Indexed: 12/22/2022] Open
Abstract
BRAF inhibitor (BRAFi) has been used for treatment of melanomas harboring V600E mutation. Despite a high initial response rate, resistance to BRAFi is inevitable. Here, we demonstrate that BRAFi-resistant (BR) melanomas are susceptible to arginine deprivation due to inability to initiate re-expression of argininosuccinate synthetase (ASS1, a key enzyme for arginine synthesis) as well as ineffective autophagy. Autophagy and ASS1 re-expression are known to protect melanoma cells from cell death upon arginine deprivation. When melanoma cells become BR cells by long-term in vitro incubation with BRAFi, c-Myc-mediated ASS1 re-expression and the levels of autophagy-associated proteins (AMPK-α1 and Atg5) are attenuated. Furthermore, our study uncovers that downregulation of deubiquitinase USP28 which results in more active c-Myc degradation via ubiquitin-proteasome machinery is the primary mechanism for inability to re-express ASS1 upon arginine deprivation in BR cells. Overexpression of USP28 in BR cells enhances c-Myc expression and hence increases ASS1 transcription upon arginine deprivation, and consequently leads to cell survival. On the other hand, overexpression of Atg5 or AMPK-α1 in BR cells can redirect arginine deprivation-induced apoptosis toward autophagy. The xenograft models also confirm that BR tumors possess lower expression of ASS1 and are hypersensitive to arginine deprivation. These biochemical changes in BRAFi resistance which make them vulnerable to arginine deprivation can be exploited for the future treatment of BR melanoma patients.
Collapse
Affiliation(s)
- Ying-Ying Li
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, Florida, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Chunjing Wu
- Division of Hematology and Oncology, Miami Veterans Affairs Healthcare System, Miami, Florida, USA
| | - Shu-Mei Chen
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.,Department of Neurosurgery, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan
| | - Sumedh S Shah
- Dauer Electron Microscopy Laboratory, Department of Biology, University of Miami, Miami, FL, USA
| | - Medhi Wangpaichitr
- Division of Hematology and Oncology, Miami Veterans Affairs Healthcare System, Miami, Florida, USA.,Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Lynn G Feun
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Macus T Kuo
- Department of Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Miguel Suarez
- Department of Laboratory Medicine, Miami Veterans Affairs Healthcare System, Miami, Florida, USA
| | - Jeffrey Prince
- Dauer Electron Microscopy Laboratory, Department of Biology, University of Miami, Miami, FL, USA
| | - Niramol Savaraj
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA.,Division of Hematology and Oncology, Miami Veterans Affairs Healthcare System, Miami, Florida, USA
| |
Collapse
|
11
|
Huang CY, Wang L, Feng CJ, Yu P, Cai XH, Yao WX, Xu Y, Liu XK, Zhu WJ, Wang Y, Zhou J, Lu Y, Wang YS. Bisphosphonates enhance EGFR-TKIs efficacy in advanced NSCLC patients with EGFR activating mutation: A retrospective study. Oncotarget 2016; 7:66480-66490. [PMID: 26624882 PMCID: PMC5341815 DOI: 10.18632/oncotarget.5515] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 09/05/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Bisphosphonates have exhibited anti-tumor activity in non-small cell lung cancer (NSCLC). We aimed to evaluate whether the combination of bisphosphonates with tyrosine kinase inhibitors of EGFR (EGFR-TKIs) could obtain a synergistic effect on advanced NSCLC patients with EGFR mutations. METHODS Between January 2008 and October 2013, 114 advanced EGFR mutations NSCLC patients who received EGFR-TKIs as first-line therapy were recruited from two cancer centers. Patients were separated into EGFR-TKIs alone or EGFR-TKIs plus bisphosphonates (combination) group. Median progression free survival (mPFS), median overall survival (mOS) distributions and survival curves were analyzed. RESULTS Among the 114 patients, 62 had bone metastases (19 patients treated with EGFR-TKIs, 43 patients treated with EGFR-TKIs + bisphosphonates). Median PFS and OS were significantly improved in combination group compared with EGFR-TKIs group (mPFS: 15.0 vs 7.3 months, P = 0.0017; mOS: 25.2 vs 10.4 months, P = 0.0015) in patients with bone metastases. Among the 71 patients (19 patients with bone metastases) treated with EGFR-TKIs alone, patients with bone metastases had poor survival prognosis (mPFS:7.3 vs 12.1 months, P = 0.0434; mOS:10.4 vs 22.0 months, P = 0.0036). The survival of patients with bone metastases who received EGFR-TKIs plus bisphosphonates therapy was non-inferior to patients without bone metastases treated with EGFR-TKIs alone (mPFS: 15.0 vs 12.1 months, p = 0.1871; mOS: 25.2 vs 22.0 months, p = 0.9798). CONCLUSIONS Concomitant use of bisphosphonates and EGFR-TKIs improves therapeutic efficacy and brings survival benefits to NSCLC patients with EGFR mutation and bone metastases.
Collapse
Affiliation(s)
- Chu-Ying Huang
- Department of Thoracic Oncology, Cancer Center, and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Medical Oncology, Enshi Tujia and Miao Autonomous Prefecture Central Hospital, Hubei Province, China
| | - Li Wang
- Department of Thoracic Oncology, Cancer Center, and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Dermatology, Enshi Tujia and Miao Autonomous Prefecture Central Hospital, Hubei Province, China
| | - Cheng-Jun Feng
- Department of Thoracic Oncology, Cancer Center, and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ping Yu
- Department of Oncology, The Second People's Hospital of Sichuan, Chengdu, China
| | - Xiao-Hong Cai
- Department of Oncology, The Second People's Hospital of Sichuan, Chengdu, China
| | - Wen-Xiu Yao
- Department of Oncology, The Second People's Hospital of Sichuan, Chengdu, China
| | - Yong Xu
- Department of Thoracic Oncology, Cancer Center, and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiao-Ke Liu
- Department of Thoracic Oncology, Cancer Center, and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wen-Jiang Zhu
- Department of Thoracic Oncology, Cancer Center, and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Wang
- Department of Thoracic Oncology, Cancer Center, and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Oncology, The First People's Hospital of Longquanyi District, Chengdu, China
| | - Jin Zhou
- Department of Oncology, The Second People's Hospital of Sichuan, Chengdu, China
| | - You Lu
- Department of Thoracic Oncology, Cancer Center, and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yong-Sheng Wang
- Department of Thoracic Oncology, Cancer Center, and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Jain RK, Hong DS, Naing A, Wheler J, Helgason T, Shi NY, Gad Y, Kurzrock R. Novel phase I study combining G1 phase, S phase, and G2/M phase cell cycle inhibitors in patients with advanced malignancies. Cell Cycle 2016; 14:3434-40. [PMID: 26467427 DOI: 10.1080/15384101.2015.1090065] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
PURPOSE Cancer is a manifestation of aberrant cellular proliferation, and the cell cycle is one of the most successfully drugged targets in oncology. No prior study has been reported that simultaneously targets the 3 principal cell cycle phases populated by proliferating cells--G1, S, and G2/M. METHODS Temsirolimus (G1 inhibitor), topotecan (S inhibitor), and bortezomib (G2/M inhibitor) were administered in combination to patients with advanced malignancies using a 3+3 dose escalation schedule to assess the safety and establish the maximum tolerated dose (primary endpoints) of this cell cycle targeting approach. An in silico pharmacodynamic model using established effects of each of these agents on the cell cycle was used to validate the regimen and to guide the dosing regimen. RESULTS Sixty-two subjects were enrolled. The most common adverse events and dose-limiting toxicities were cytopenias, consistent with the cell cycle targeting approach employed. All cytopenias resolved to baseline values upon holding study drug administration. The maximum tolerated dose was temsirolimus 15 mg/kg IV D1, 8, 15; topotecan 2.8 mg/m(2) IV D1, 8; and bortezomib 0.6 mg/m2 IV D1, 4, 8, 11 [DOSAGE ERROR CORRECTED] of a 21-day cycle. In silico modeling suggests the regimen induces cell population shifts from G2/M and S phases to G1 phase and the quiescent G0 phase. Eighteen percent of subjects (11/62) achieved partial response (n = 2, serous ovarian and papillary thyroid) or stable disease for > 6 months (n = 9). CONCLUSION Combining drugs with inhibitory activity of G1 phase, S phase, and G2/M phase is safe and warrants further evaluation.
Collapse
Affiliation(s)
- Rajul K Jain
- a Department of Investigational Cancer Therapeutics (Phase I Program) ; MD Anderson Cancer Center ; Houston , TX USA
| | - David S Hong
- a Department of Investigational Cancer Therapeutics (Phase I Program) ; MD Anderson Cancer Center ; Houston , TX USA
| | - Aung Naing
- a Department of Investigational Cancer Therapeutics (Phase I Program) ; MD Anderson Cancer Center ; Houston , TX USA
| | - Jennifer Wheler
- a Department of Investigational Cancer Therapeutics (Phase I Program) ; MD Anderson Cancer Center ; Houston , TX USA
| | - Thorunn Helgason
- a Department of Investigational Cancer Therapeutics (Phase I Program) ; MD Anderson Cancer Center ; Houston , TX USA
| | - Nai-Yi Shi
- a Department of Investigational Cancer Therapeutics (Phase I Program) ; MD Anderson Cancer Center ; Houston , TX USA
| | | | - Razelle Kurzrock
- c Moores Cancer Center; University of California San Diego ; La Jolla , CA USA
| |
Collapse
|
13
|
Feng C, Liu X, Li X, Guo F, Huang C, Qin Q, Wang Y. Zoledronic acid increases the antitumor effect of gefitinib treatment for non-small cell lung cancer with EGFR mutations. Oncol Rep 2016; 35:3460-70. [PMID: 27109760 DOI: 10.3892/or.2016.4741] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 10/31/2015] [Indexed: 02/05/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) patients with epithelial growth factor receptor (EGFR) mutations and bone metastases are often concurrently administered tyrosine kinase inhibitors (TKIs) and bisphosphonates. Yet, the effects and mechanisms of these agents are unclear. In the present study, we aimed to ascertain whether zoledronic acid (ZA) increases the antitumor effects of gefitinib treatment on NSCLC with EGFR mutations and the related mechanisms of action. The effects of ZA and gefitinib on NSCLC tumor cells with EGFR mutations (HCC827, HCC827 GR and H1975) in regards to proliferation, apoptosis, cell cycle and signaling pathways were detected. ZA increased the antitumor effects of gefitinib on NSCLC with EGFR activating mutations and TKI resistance in vitro. Gefitinib caused cell cycle arrest in the G0/G1 phase, ZA induced S phase accumulation and the effect of the combined treatment was neutralization. Combined treatment obviously inhibited STAT3 and/or p‑STAT3 protein expression compared with treatment with each single drug in vitro and in vivo, and it also significantly inhibited TKI resistance NSCLC tumor growth in vivo. In conclusion, ZA increased the antitumor effects of gefitinib on NSCLC with EGFR activating mutations and TKI resistance by regulating the cell cycle, inducing caspase-3 expression and inhibiting STAT3 expression.
Collapse
Affiliation(s)
- Chengjun Feng
- Department of Thoracic Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaoke Liu
- Department of Thoracic Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaoyu Li
- Department of Thoracic Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Fuchun Guo
- Department of Thoracic Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Chuying Huang
- Department of Thoracic Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qing Qin
- Department of Thoracic Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yongsheng Wang
- Department of Thoracic Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
14
|
Biray Avci C, Kurt CC, Tepedelen BE, Ozalp O, Goker B, Mutlu Z, Dodurga Y, Elmas L, Gunduz C. Zoledronic acid induces apoptosis via stimulating the expressions of ERN1, TLR2, and IRF5 genes in glioma cells. Tumour Biol 2015; 37:6673-9. [PMID: 26646564 DOI: 10.1007/s13277-015-4519-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/25/2015] [Indexed: 11/28/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive brain tumor that affects older people. Although the current therapeutic approaches for GBM include surgical resection, radiotherapy, and chemotherapeutic agent temozolomide, the median survival of patients is 14.6 months because of its aggressiveness. Zoledronic acid (ZA) is a nitrogen-containing bisphosphonate that exhibited anticancer activity in different cancers. The purpose of this study was to assess the potential effect of ZA in distinct signal transduction pathways in U87-MG cells. In this study, experiments performed on U87-MG cell line (Human glioblastoma-astrocytoma, epithelial-like cell line) which is an in vitro model of human glioblastoma cells to examine the cytotoxic and apoptotic effects of ZA. IC50 dose of ZA, 25 μM, applied on U87-MG cells during 72 h. ApoDIRECT In Situ DNA Fragmentation Assay was used to investigate apoptosis of U87MG cells. The quantitative reverse transcription polymerase chain reaction (qRT-PCR) (LightCycler480 System) was carried out for 48 gene expression like NF-κB, Toll-like receptors, cytokines, and inteferons. Our results indicated that ZA (IC50 dose) increased apoptosis 1.27-fold in U87MG cells according to control cells. According to qRT-PCR data, expression levels of the endoplasmic reticulum-nuclei-1 (ERN1), Toll-like receptor 2 (TLR2), and human IFN regulatory factor 5 (IRF5) tumor suppressor genes elevated 2.05-, 2.08-, and 2.3-fold by ZA, respectively, in U87MG cells. Our recent results indicated that ZA have a key role in GBM progression and might be considered as a potential agent in glioma treatment.
Collapse
Affiliation(s)
- Cigir Biray Avci
- Medical Biology Department, Ege University Medical School, Bornova, Izmir, Turkey.
| | - Cansu Caliskan Kurt
- Medical Biology Department, Ege University Medical School, Bornova, Izmir, Turkey
| | - Burcu Erbaykent Tepedelen
- Department of Molecular Biology and Genetics, Avrasya University Faculty of Science and Letters, Trabzon, Turkey
| | - Ozgun Ozalp
- Medical Biology Department, Ege University Medical School, Bornova, Izmir, Turkey
| | - Bakiye Goker
- Medical Biology Department, Ege University Medical School, Bornova, Izmir, Turkey
| | - Zeynep Mutlu
- Medical Biology Department, Ege University Medical School, Bornova, Izmir, Turkey
| | - Yavuz Dodurga
- Medical Biology Department, Pamukkale University Medical School, Denizli, Turkey
| | - Levent Elmas
- Medical Biology Department, Pamukkale University Medical School, Denizli, Turkey
| | - Cumhur Gunduz
- Medical Biology Department, Ege University Medical School, Bornova, Izmir, Turkey
| |
Collapse
|
15
|
Ge XY, Yang LQ, Jiang Y, Yang WW, Fu J, Li SL. Reactive oxygen species and autophagy associated apoptosis and limitation of clonogenic survival induced by zoledronic acid in salivary adenoid cystic carcinoma cell line SACC-83. PLoS One 2014; 9:e101207. [PMID: 24963720 PMCID: PMC4071064 DOI: 10.1371/journal.pone.0101207] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Accepted: 06/04/2014] [Indexed: 01/10/2023] Open
Abstract
Salivary adenoid cystic carcinoma is an epithelial tumor in the head and neck region. Despite its slow growth, patients with salivary adenoid cystic carcinoma exhibit poor long term survival because of a high rate of distant metastasis. Lung and bone are common distant metastasis sites. Zoledronic acid, a third generation bisphosphonate, has been used for tumor-induced osteolysis due to bone metastasis and has direct antitumor activity in several human neoplasms. Here, we observed that zoledronic acid inhibited salivary adenoid cystic carcinoma cell line SACC-83 xenograft tumor growth in nude mice. In vitro, zoledronic acid induced apoptosis and reduced clonogenic survival in SACC-83. Flow cytometry and western blotting indicated that the cell cycle was arrested at G0/G1. Zoledronic acid treatment upregulated reactive oxygen species as well as the autophagy marker protein LC-3B. Reactive oxygen species scavenger N-acetylcysteine and autophagy antagonist 3-methyladenine decreased zoledronic acid-induced apoptosis and increased clonogenic survival. Silencing of the autophagy related gene Beclin-1 also decreased zoledronic acid-induced apoptosis and inhibition of clonogenic formation. In addition, isobolographic analysis revealed synergistic effects on apoptosis when zoledronic acid and paclitaxel/cisplatin were combined. Taken together, our results suggest that zoledronic acid induced apoptosis and reduced clonogenic survival via upregulation of reactive oxygen species and autophagy in the SACC-83 cell line. Thus, zoledronic acid should be considered a promising drug for the treatment of salivary adenoid cystic carcinoma.
Collapse
Affiliation(s)
- Xi-Yuan Ge
- Central Laboratory, Peking University School and Hospital of Stomatology, Haidian District, Beijing, PR China
| | - Lin-Qian Yang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Haidian District, Beijing, PR China
| | - Yang Jiang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Haidian District, Beijing, PR China
| | - Wen-Wen Yang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Haidian District, Beijing, PR China
| | - Jia Fu
- Central Laboratory, Peking University School and Hospital of Stomatology, Haidian District, Beijing, PR China
| | - Sheng-Lin Li
- Central Laboratory, Peking University School and Hospital of Stomatology, Haidian District, Beijing, PR China
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Haidian District, Beijing, PR China
- * E-mail:
| |
Collapse
|
16
|
Hirsh V. Targeted treatments of bone metastases in patients with lung cancer. Front Oncol 2014; 4:146. [PMID: 24982847 PMCID: PMC4058774 DOI: 10.3389/fonc.2014.00146] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 05/28/2014] [Indexed: 11/17/2022] Open
Abstract
Until now ~30–40% of patients with advanced lung cancer develop bone metastases, but as the newer therapies are extending survival, the chance of developing bone metastases increases. Bone metastases cause skeletal-related events (SREs) such as pathologic fractures, spinal cord compression, radiation therapy or surgery to bone, or hypercalcemia, which can have debilitating consequences affecting patients’ health-related quality of life (HR-QOL) and performance status (PS). Poor PS then prevents the patients to receive further lines of treatments, which are available today. SREs are associated with increased economic costs. In one clinical trial, the median time to first SRE was only 5 months. Early detection of bone metastases can prevent SREs and avoid inappropriate implementation of major surgery or chemoradiation therapy. With the new generation bisphosphonate zoledronic acid (ZA) or denosumab (anti-RANKL activity), one can reduce the number of patients who experience SREs, decrease the annual incidence of SREs and delay the median time to first SRE. These agents are effective even after the onset of SREs. They are well tolerated, with manageable side effects. The biochemical markers of bone metabolism especially N-telopeptide of type I collagen and bone specific alkaline phosphatase (BALP) can be both prognostic and predictive markers for the patients with bone metastases from non-small cell lung cancer (NSCLC). Anticancer activity of ZA and denosumab further supports their use as soon as bone metastases are diagnosed in patients with NSCLC. Further trials will inform us about the efficacy of these agents for prevention of bone metastases and even about possible effects on visceral metastases.
Collapse
Affiliation(s)
- Vera Hirsh
- McGill University Health Centre, Royal Victoria Hospital , Montreal, QC , Canada
| |
Collapse
|
17
|
Abstract
Cancer risk reduction using pharmacological means is an attractive modern preventive approach that supplements the classical behavioural prevention recommendations. Medications that are commonly used by large populations to treat a variety of common, non-cancer-related, medical situations are an attractive candidate pool. This Review discusses three pharmacological agents with the most evidence for their potential as cancer chemopreventive agents: anti-hypercholesterolaemia medications (statins), an antidiabetic agent (metformin) and antiosteoporosis drugs (bisphosphonates). Data are accumulating to support a significant negative association of certain statins with cancer occurrence or survival in several major tumour sites (mostly gastrointestinal tumours and breast cancer), with an augmented combined effect with aspirin or other non-steroidal anti-inflammatory drugs. Metformin, but not other hypoglycaemic drugs, also seems to have some antitumour growth activity, but the amount of evidence in human studies, mainly in breast cancer, is still limited. Experimental and observational data have identified bisphosphonates as a pharmacological group that could have significant impact on incidence and mortality of more than one subsite of malignancy. At the current level of evidence these potential chemopreventive drugs should be considered in high-risk situations or using the personalized approach of maximizing individual benefits and minimizing the potential for adverse effects with the aid of pharmacogenetic indicators.
Collapse
|
18
|
Insalaco L, Di Gaudio F, Terrasi M, Amodeo V, Caruso S, Corsini LR, Fanale D, Margarese N, Santini D, Bazan V, Russo A. Analysis of molecular mechanisms and anti-tumoural effects of zoledronic acid in breast cancer cells. J Cell Mol Med 2013; 16:2186-95. [PMID: 22260151 PMCID: PMC3822988 DOI: 10.1111/j.1582-4934.2012.01527.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Zoledronic acid (ZOL) is the most potent nitrogen-containing bisphosphonate (N-BPs) that strongly binds to bone mineral and acts as a powerful inhibitor of bone resorption, already clinically available for the treatment of patients with osteolytic metastases. Recent data also suggest that ZOL, used in breast cancer, may provide more than just supportive care modifying the course of the disease, though the possible molecular mechanism of action is still unclear.As breast cancer is one of the primary tumours with high propensity to metastasize to the bone, we investigated, for the first time, differential gene expression profile on Michigan Cancer Foundation-7 (MCF-7) breast cancer cells treated with low doses of ZOL (10 μM). Microarrays analysis was used to identify, describe and summarize evidence regarding the molecular basis of actions of ZOL and of their possible direct anti-tumour effects. We validated gene expression results of specific transcripts involved in major cellular process by Real Time and Western Blot analysis and we observed inhibition of proliferation and migration through 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and Matrigel assay. We then focused on changes in the cytoskeletal components as fibronectin 1 (FN1), actin, and anti angiogenic compounds as transforming growth factor-β1 (TGF-β1) and thrombospondin 1 (THBS1). The up-regulation of these products may have an important role in inhibiting proliferation, invasion and angiogenesis mediated by ZOL.
Collapse
Affiliation(s)
- Lavinia Insalaco
- Section of Medical Oncology, Department of Surgical and Oncology, University of Palermo, Palermo, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Li XY, Lin YC, Huang WL, Lin W, Wang HB, Lin WZ, Lin SL. Zoledronic acid inhibits human nasopharyngeal carcinoma cell proliferation by activating mitochondrial apoptotic pathway. Med Oncol 2012; 29:3374-80. [PMID: 22729369 DOI: 10.1007/s12032-012-0281-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 06/08/2012] [Indexed: 02/05/2023]
Abstract
We investigated the apoptosis-inducing effect of zoledronic acid in human nasopharyngeal carcinoma cell HNE-1 and explore the potential mechanism. Human nasopharyngeal carcinoma cell HNE-1 was exposed to various concentrations (0-40 μmol/L) of zoledronic acid. Cell proliferation was studied by an MTT assay. Cell apoptosis was analyzed by flow cytometry and TdT-mediated dUTP nick-end labeling (TUNEL) assay. Cell cycle was analyzed by flow cytometry. Gene expressions were investigated by quantitative real-time PCR, and protein expressions were investigated by Western blot. The results showed zoledronic acid decreased cell proliferation not in a time- or dose-dependent fashion. TUNEL assay, together with Annexin V/propidium iodide FACS analysis, confirmed the increase in apoptotic HNE-1 cells treated with zoledronic acid. Cell cycle analysis showed a larger number of treated cells occupied the S-phase. Quantitative RT-PCR and Western blot revealed that the pro-apoptotic genes, Bad, Bax, and Caspase-9, were upregulated in treated HNE-1 cells, whereas the anti-apoptotic gene, Bcl-2, was downregulated in both mRNA and protein levels. In conclusion, zoledronic inhibits human nasopharyngeal carcinoma cell proliferation by inducing apoptosis via the mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Xu-Yuan Li
- Department of Internal Medicine, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, 515031, China
| | | | | | | | | | | | | |
Collapse
|
20
|
Zoledronic acid induces S-phase arrest via a DNA damage response in normal human oral keratinocytes. Arch Oral Biol 2012; 57:906-17. [DOI: 10.1016/j.archoralbio.2011.11.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 11/11/2011] [Accepted: 11/20/2011] [Indexed: 01/16/2023]
|
21
|
Li XY, Lin YC, Huang WL, Hong CQ, Chen JY, You YJ, Li WB. Zoledronic acid inhibits proliferation and impairs migration and invasion through downregulating VEGF and MMPs expression in human nasopharyngeal carcinoma cells. Med Oncol 2012; 29:714-20. [PMID: 21431960 DOI: 10.1007/s12032-011-9904-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 03/08/2011] [Indexed: 02/05/2023]
Abstract
To demonstrate the effect of zoledronic acid in proliferation, invasion, and migration of human nasopharyngeal carcinoma cell HNE-1 and explore the potential role of VEGF, MMP-2, and MMP-9 proteins in vitro. Human nasopharyngeal carcinoma cell HNE-1 was exposed to various concentrations (0-40 μmol/l) of zoledronic acid. Zoledronic acid inhibited proliferation of HNE-1 cells though not in a dose-dependent manner. Zoledronic acid had exerted a dose-dependent effect on the migration and invasion of HNE-1 cells. Both expressions of mRNA and protein of MMP2, MMP9, and VEGF were reduced, respectively, detected by RT-PCR and Western blot assays. These data suggested that zoledronic acid not only inhibited growth but also invasion and migration of HNE-1 cells in vitro. The anti-cancer action of zoledronic acid was partially associated with the suppression of VEGF expression and secretion and downregulating the expression of MMP2 and MMP9.
Collapse
Affiliation(s)
- Xu-Yuan Li
- Department of Internal Medicine, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou 515031, China
| | | | | | | | | | | | | |
Collapse
|
22
|
Mani J, Vallo S, Barth K, Makarević J, Juengel E, Bartsch G, Wiesner C, Haferkamp A, Blaheta RA. Zoledronic acid influences growth, migration and invasive activity of prostate cancer cells in vitro. Prostate Cancer Prostatic Dis 2012; 15:250-5. [DOI: 10.1038/pcan.2012.9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
23
|
Henk HJ, Kaura S, Teitelbaum A. Retrospective evaluation of the clinical benefit of long-term continuous use of zoledronic acid in patients with lung cancer and bone metastases. J Med Econ 2012; 15:195-204. [PMID: 22175657 DOI: 10.3111/13696998.2011.650489] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND For patients with bone metastases, skeletal-related events including fracture are common, can cause considerable morbidity, and may reduce overall survival (OS). This retrospective analysis assessed the effect of Zometa (zoledronic acid, ZOL), an intravenous bisphosphonate (IV-BP), on fracture risk and OS in patients with bone metastases from lung cancer (LC). (Zometa is a registered trademark of Novartis Pharmaceuticals Corporation, USA.) METHODS A claims-based analysis using commercial and Medicare Advantage data from >45 US managed-care plans was used to evaluate the association between fracture risk and treatment persistency (31-90, 91-180, 181-365, and ≥366 days) and follow-up duration in LC patients diagnosed with bone metastases between 01/01/2001 and 12/31/2006 and treated with ZOL or without (no IV-BP). Persistency was defined as the absence of a >45-day gap between ZOL treatments. Analysis of variance tests were used to compare follow-up duration, a proxy for OS, between ZOL persistency groups. The effect of time to treatment with ZOL was also assessed. RESULTS In 9874 LC patients with bone metastases (n = 1090 ZOL; n = 8784 no IV-BP) the unadjusted relative fracture risk was reduced by 40% with ZOL vs no IV-BP; fracture risk decreased consistently with increasing duration of ZOL treatment. Even short-term (31-90 days) ZOL significantly reduced fracture risk (47%) vs no IV-BP (p = 0.005) with adjustment for differences in demographic and clinical characteristics. Delaying ZOL until after bone metastases were diagnosed significantly increased fracture risk (p = 0.0017). For a sub-set of patients included in a survival analysis (n = 550 ZOL; n = 4512 no IV-BP), mortality was significantly lower (mean, 38.6 vs 46.8 deaths/100 person-years; p = 0.038) in those treated with ZOL vs no IV-BP. LIMITATIONS Interpretation of this claims-based analysis must be tempered by the inherent limitations of observational data, such as limited clinical information and the ability to control for prognostic factors. CONCLUSIONS This retrospective analysis demonstrates that LC patients with bone metastases receiving ZOL had significantly reduced risk of fracture (p = 0.005) and death (p < 0.038) vs patients receiving no IV-BP. Longer ZOL persistency consistently yielded better outcomes, with ≥12 months' treatment producing the greatest benefit.
Collapse
Affiliation(s)
- Henry J Henk
- OptumInsight, Health Economics and Outcomes Research, Eden Prairie, MN 55344, USA.
| | | | | |
Collapse
|
24
|
Michailidou M, Holen I. Combinations of bisphosphonates and classical anticancer drugs: a preclinical perspective. Recent Results Cancer Res 2012; 192:145-69. [PMID: 22307374 DOI: 10.1007/978-3-642-21892-7_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bone metastases are frequent complications in advanced breast and prostate cancer among others, resulting in increased risk of fractures, pain, hypercalcaemia of malignancy and a reduction in patient independence and mobility. Bisphosphonates (BPs) are in wide clinical use for the treatment of cancer-induced bone disease associated with advanced cancer, due to their potent ability to reduce skeletal-related events (SREs) and improve quality of life. Despite the profound effect on bone health, the majority of clinical studies have failed to demonstrate an overall survival benefit of BP therapy. There is increasing preclinical evidence to suggest that inclusion of the most potent nitrogen-containing BPs (NBPs) in combination therapy results in increased antitumour effects and improved survival, but that the particular schedules used are of key importance to achieve optimal benefit. Recent clinical data have suggested that there may be effects of adjuvant NBP therapy on breast tumours outside the skeleton. These findings have led to renewed interest in the use of BPs in cancer therapy, in particular how they can be included as part of adjuvant protocols. Here we review the key data reported from preclinical model systems investigating the effects of combination therapy including BPs with particular emphasis on breast and prostate cancer.
Collapse
|
25
|
Webster MR, Zhao M, Rudek MA, Hann CL, Freel Meyers CL. Bisphosphonamidate clodronate prodrug exhibits potent anticancer activity in non-small-cell lung cancer cells. J Med Chem 2011; 54:6647-56. [PMID: 21863853 DOI: 10.1021/jm200521a] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Bisphoshonates are used clinically to treat disorders of calcium metabolism, hypercalcemia and osteoporosis, and malignant bone disease. Although these agents are commonly used in cancer patients and have potential direct anticancer effects, their use for the treatment of extraskeletal disease is limited as a result of poor cellular uptake. We have designed and synthesized bisphosphonamidate prodrugs that undergo intracellular activation to release the corresponding bisphosphonate and require only two enzymatic activation events to unmask multiple negative charges. We demonstrate efficient bisphosphonamidate activation and significant enhancement in anticancer activity of two bisphosphonamidate prodrugs in vitro compared to the parent bisphosphonate. These data suggest a novel approach to optimizing the anticancer activities of commonly used bisphosphonates.
Collapse
Affiliation(s)
- Marie R Webster
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, WBSB 305, Baltimore, Maryland 21201, United States
| | | | | | | | | |
Collapse
|
26
|
Brown HK, Ottewell PD, Coleman RE, Holen I. The kinetochore protein Cenp-F is a potential novel target for zoledronic acid in breast cancer cells. J Cell Mol Med 2011; 15:501-13. [PMID: 20015195 PMCID: PMC3922372 DOI: 10.1111/j.1582-4934.2009.00995.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The anti-resorptive agent zoledronic acid inhibits key enzymes in the mevalonate pathway, disrupting post-translational modification and thereby correct protein localization and function. Inhibition of prenylation may also be responsible for the reported anti-tumour effects of zoledronic acid, but the specific molecular targets have not been identified. Cenp-F/mitosin, a kinetochore-associated protein involved in the correct separation of chromosomes during mitosis, has been shown to undergo post-translational prenylation and may therefore be a novel target contributing to the anti-tumour effects of zoledronic acid. We investigated whether zoledronic acid causes loss of Cenp-F from the kinetochore in breast cancer cells, to determine if the reported anti-tumour effects may be mediated by impairing correct chromosome separation. MDA-MB-436, MDA-MB-231 and MCF-7 breast cancer cells and MCF-10A non-malignant breast epithelial cells were treated with zoledronic acid in vitro, and the effect on Cenp-F localization was analysed by immunoflourescence microscopy. Zoledronic acid caused loss of Cenp-F from the kinetochore, accompanied by an increase in the number of cells in pro-, /prometa- and metaphase in all of the cancer cell lines. There was also a significant increase in the number of lagging chromosomes in mitotic cells. The effects of zoledronic acid could be reversed by inclusion of an intermediary of the mevalonate pathway, showing that the loss of Cenp-F from the kinetochore was caused by the inhibition of farnesylation. In contrast, no effect was seen on Cenp-F in non-malignant MCF-10A cells. This is the first report showing a specific effect of zoledronic acid on a protein involved in the regulation of chromosome segregation, identifying Cenp-F as a potential new molecular target for NBPs in tumour cells.
Collapse
Affiliation(s)
- Hannah K Brown
- Academic Unit of Clinical Oncology, Medical School, University of Sheffield, Sheffield, UK
| | | | | | | |
Collapse
|
27
|
Ressler S, Mlineritsch B, Greil R. Zoledronic acid for adjuvant use in patients with breast cancer. Expert Rev Anticancer Ther 2011; 11:333-49. [PMID: 21417849 DOI: 10.1586/era.11.13] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Zoledronic acid, a nitrogen-containing bisphosphonate, is firmly established in the management of metastatic bone disease. It inhibits farnesyl diphosphonate synthase within the mevalonate pathway and, through this mechanism, is a potent inhibitor of osteoclast-mediated bone resorption. In addition, there are preclinical data suggesting that farnesyl diphosphonate synthase inhibition by zoledronic acid has anti-tumor effects in breast cancer. Adjuvant therapies for early breast cancer are associated with substantial decreases in bone mineral density. Results from three clinical trials, ABCSG-12, Z-FAST and ZO-FAST, indicate that the addition of twice-yearly zoledronic acid to standard adjuvant endocrine therapy in premenopausal and postmenopausal patients with hormone receptor-positive breast cancer prevents cancer treatment-induced bone loss. Moreover, it is becoming evident that it may also exert anticancer effects in an estrogen-deprived state in the adjuvant and neoadjuvant setting. However, long-term side effects need to be taken into consideration for treatment decisions.
Collapse
Affiliation(s)
- Sigrun Ressler
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory of Immunological and Molecular Cancer Research, Private Medical University Hospital, Salzburg, Austria
| | | | | |
Collapse
|
28
|
Aft R, Perez JR, Raje N, Hirsh V, Saad F. Could targeting bone delay cancer progression? Potential mechanisms of action of bisphosphonates. Crit Rev Oncol Hematol 2011; 82:233-48. [PMID: 21683613 DOI: 10.1016/j.critrevonc.2011.05.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 05/18/2011] [Accepted: 05/25/2011] [Indexed: 12/12/2022] Open
Abstract
Although dissemination may occur early in the course of many cancers, the development of overt metastases depends upon a variety of factors inherent to the cancer cells and the tissue(s) they colonize. The time lag between initial dissemination and established metastases could be several years, during which period the bone marrow may provide an unwitting sanctuary for disseminated tumor cells (DTCs). Survival in a dormant state within the bone marrow may help DTCs weather the effects of anticancer therapies and seed posttreatment relapses. The importance of the bone marrow for facilitating DTC survival may vary depending on the type of cancer and mechanisms of tumor cell dissemination. By altering the bone microenvironment, bisphosphonates may reduce DTC viability. Moreover, some bisphosphonates have demonstrated multiple anticancer activities. These multiple mechanisms may help explain the improvement in disease outcomes with the use of zoledronic acid in malignancies like breast cancer and multiple myeloma.
Collapse
Affiliation(s)
- Rebecca Aft
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | | | | | | | | |
Collapse
|
29
|
Li X, Liao J, Park SI, Koh AJ, Sadler WD, Pienta KJ, Rosol TJ, McCauley LK. Drugs which inhibit osteoclast function suppress tumor growth through calcium reduction in bone. Bone 2011; 48:1354-61. [PMID: 21419883 PMCID: PMC3457787 DOI: 10.1016/j.bone.2011.03.687] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 03/10/2011] [Accepted: 03/11/2011] [Indexed: 11/26/2022]
Abstract
Prostate carcinoma frequently metastasizes to bone where the microenvironment facilitates its growth. Inhibition of bone resorption is effective in reducing tumor burden and bone destruction in prostate cancer. However, whether drugs that inhibit osteoclast function inhibit tumor growth independent of inhibition of bone resorption is unclear. Calcium is released during bone resorption and the calcium sensing receptor is an important regulator of cancer cell proliferation. The goal of this investigation was to elucidate the role of calcium released during bone resorption and to determine the impact of drugs which suppress bone resorption on tumor growth in bone. To compare tumor growth in a skeletal versus non-skeletal site, equal numbers of canine prostate cancer cells expressing luciferase (ACE-1(luc)) were inoculated into a simple collagen matrix, neonatal mouse vertebrae (vossicles), human de-proteinized bone, or a mineralized collagen matrix. Implants were placed subcutaneously into athymic mice. Luciferase activity was used to track tumor growth weekly, and at one month tumors were dissected for histologic analysis. Luciferase activity and tumor size were greater in vossicles, de-proteinized bone and mineralized collagen matrix versus non-mineralized collagen implants. The human osteoblastic prostate carcinoma cell line C4-2b also grew better in a mineral rich environment with a greater proliferation of C4-2b cells reflected by Ki-67 staining. Zoledronic acid (ZA), a bisphosphonate, and recombinant OPG-Fc, a RANKL inhibitor, were administered to mice bearing vertebral implants (vossicles) containing ACE-1 osteoblastic prostate cancer cells. Vossicles or collagen matrices were seeded with ACE-1(luc) cells subcutaneously in athymic mice (2 vossicles, 2 collagen implants/mouse). Mice received ZA (5 μg/mouse, twice/week), (OPG-Fc at 10mg/kg, 3 times/week) or vehicle, and luciferase activity was measured weekly. Histologic analysis of the tumors, vossicles and endogenous bones and serum biochemistry were performed. Antiresorptive administration was associated with decreased serum TRAP5b, reduced osteoclast numbers, and increased tibia and vossicle bone areas. ZA significantly decreased bone marrow calcium concentrations without affecting serum calcium. ZA and OPG-Fc significantly inhibited tumor growth in bone but not in collagen implants. In conclusion, the inhibitory effects of ZA or OPG-Fc on prostate tumor growth in bone are mediated via blocking bone resorption and calcium release from bone.
Collapse
Affiliation(s)
- Xin Li
- Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Singh SK, Baumgart S, Singh G, König AO, Reutlinger K, Hofbauer LC, Barth P, Gress TM, Lomberk G, Urrutia R, Fernandez-Zapico ME, Ellenrieder V. Disruption of a nuclear NFATc2 protein stabilization loop confers breast and pancreatic cancer growth suppression by zoledronic acid. J Biol Chem 2011; 286:28761-28771. [PMID: 21628454 DOI: 10.1074/jbc.m110.197533] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The aminobisphosphonate zoledronic acid has elicited significant attention due to its remarkable anti-tumoral activity, although its detailed mechanism of action remains unclear. Here, we demonstrate the existence of a nuclear GSK-3β-NFATc2 stabilization pathway that promotes breast and pancreatic cancer growth in vitro and in vivo and serves as a bona fide target of zoledronic acid. Specifically, the serine/threonine kinase GSK-3β stabilizes nuclear NFATc2 through phosphorylation of the serine-rich SP2 domain, thus protecting the transcription factor from E3-ubiquitin ligase HDM2-mediated proteolysis. Zoledronic acid disrupts this NFATc2 stabilization pathway through two mechanisms, namely GSK-3β inhibition and induction of HDM2 activity. Upon nuclear accumulation, HDM2 targets unphosphorylated NFATc2 for ubiquitination at acceptor lysine residues Lys-684/Lys-897 and hence labels the factor for subsequent proteasomal degradation. Conversely, mutagenesis-induced constitutive serine phosphorylation (Ser-215, Ser-219, and Ser-223) of the SP2 domain prevents NFATc2 from HDM2-mediated ubiquitination and degradation and consequently rescues cancer cells from growth suppression by zoledronic acid. In conclusion, this study demonstrates a critical role of the GSK-3β-HDM2 signaling loop in the regulation of NFATc2 protein stability and growth promotion and suggests that double targeting of this pathway is responsible, at least to a significant part, for the potent and reliable anti-tumoral effects of zoledronic acid.
Collapse
Affiliation(s)
- Shiv K Singh
- Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps-University of Marburg, D-35043 Marburg, Germany
| | - Sandra Baumgart
- Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps-University of Marburg, D-35043 Marburg, Germany
| | - Garima Singh
- Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps-University of Marburg, D-35043 Marburg, Germany
| | - Alexander O König
- Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps-University of Marburg, D-35043 Marburg, Germany
| | - Kristina Reutlinger
- Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps-University of Marburg, D-35043 Marburg, Germany
| | - Lorenz C Hofbauer
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technical University, D-01307 Dresden, Germany
| | - Peter Barth
- Department of Pathology, Philipps-University of Marburg, D-35043 Marburg, Germany
| | - Thomas M Gress
- Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps-University of Marburg, D-35043 Marburg, Germany
| | - Gwen Lomberk
- Laboratory of Epigenetics and Chromatin Dynamics, Departments of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, and
| | - Raul Urrutia
- Laboratory of Epigenetics and Chromatin Dynamics, Departments of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, and
| | - Martin E Fernandez-Zapico
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, Minnesota 55904
| | - Volker Ellenrieder
- Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps-University of Marburg, D-35043 Marburg, Germany,.
| |
Collapse
|
31
|
Di Salvatore M, Orlandi A, Bagalà C, Quirino M, Cassano A, Astone A, Barone C. Anti-tumour and anti-angiogenetic effects of zoledronic acid on human non-small-cell lung cancer cell line. Cell Prolif 2011; 44:139-46. [PMID: 21401755 DOI: 10.1111/j.1365-2184.2011.00745.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVES Although emerging data suggest that zoledronic acid (Zol) may have different anti-tumour activities against a broad range of cancers, its effects on lung cancer remain largely unknown. The aim of this study was to evaluate in vitro the anti-tumoural and anti-angiogenetic effect of zoledronic acid in non-small-cell lung cancer (NSCLC) cells. MATERIAL AND METHODS We treated A549 NSCLC cells with zoledronic acid to investigate survival, cell cycle activity, anti-angiogenic activity and apoptotic responses to it. RESULTS We observed that highest Zol concentration (100μm) caused arrest in G1 phase of the cell cycle and also induced different percentages of apoptosis in presence (0.9% versus 4.4%) or absence (2.4% versus 28.5%) of serum (P=0.0001). Zol concentration from 5 to 100μm for 2 days induced significant concentration-dependent cell death in adherent cells. Furthermore, Zol (10-100μm) induced dose-dependent reduction both of mRNA and protein expression of VEGF associated with parallel decrease in VEGF secretion in the culture medium. CONCLUSION Taken together, these results support a possible anti-cancer and anti-angiogenetic activity of Zol. Our data may not only provide a basis for the clinical use of this drug as preventive agent of bone metastases but also suggest that Zol deserves attention as an anti-cancer agent in non-small-cell lung cancer.
Collapse
Affiliation(s)
- M Di Salvatore
- Division of Medical Oncology, Catholic University of Sacred Heart, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
32
|
Mahtani R, Khan R, Jahanzeb M. The potential application of zoledronic acid as anticancer therapy in patients with non-small-cell lung cancer. Clin Lung Cancer 2011; 12:26-32. [PMID: 21273176 DOI: 10.3816/clc.2011.n.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Non-small-cell lung cancer (NSCLC) is frequently characterized by metastases to bone. Bisphosphonates have demonstrated efficacy in reducing the risk of skeletal-related events in cancer patients with bone metastases, including those with NSCLC. Zoledronic acid (ZA) is one of the most potent bisphosphonates and is approved for the first-line treatment of patients with multiple myeloma and bone metastases from solid tumors. Recent preclinical and clinical data suggest that ZA may also have direct and indirect anticancer effects. Several preclinical studies have provided insight into the potential mechanisms responsible for the anticancer activity of ZA, including inhibiting farnesyl pyrophosphate or geranylgeranyl pyrophosphate and activation of immune-mediated anticancer response by γδ T cells. In patients with NSCLC, ZA has been shown to reduce vascular endothelial growth factor levels with a direct correlation to clinical response. Clinical studies in this setting have shown that ZA may also provide a survival benefit and prolong time to progression. Ongoing studies are evaluating the efficacy of ZA for anticancer activity and prevention of bone metastases. Bisphosphonates, particularly ZA, are generally well tolerated and may likely offer an adjunct therapeutic option for patients with NSCLC.
Collapse
Affiliation(s)
- Reshma Mahtani
- Division of Hematology/Oncology, Sylvester Comprehensive Cancer Center, University of Miami Health Systems, Deerfield Beach, FL 33442, USA.
| | | | | |
Collapse
|
33
|
Clézardin P. Bisphosphonates' antitumor activity: an unravelled side of a multifaceted drug class. Bone 2011; 48:71-9. [PMID: 20655399 DOI: 10.1016/j.bone.2010.07.016] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2010] [Revised: 07/11/2010] [Accepted: 07/12/2010] [Indexed: 12/13/2022]
Abstract
Bisphosphonates, especially nitrogen-containing bisphosphonates (N-BPs), are widely used to preserve and improve bone health in patients with cancer because they inhibit osteoclast-mediated bone resorption. In addition to their effects on bone, preclinical evidence strongly suggests that N-BPs exert anticancer activity without the involvement of osteoclasts by interacting with macrophages, endothelial cells and tumor cells, and by stimulating the cytotoxicity of γδ T cells, a subset of human T cells. This review examines the current insights and fronts of ongoing preclinical research on N-BPs' antitumor activity.
Collapse
Affiliation(s)
- Philippe Clézardin
- INSERM, Research Unit U664, University of Lyon-1, Faculty of Medicine Lyon-Est (domaine Laennec), rue Guillaume Paradin, 69372 Lyon Cedex 08, France.
| |
Collapse
|
34
|
Abstract
Zoledronic acid effectively reduces/delays skeletal-related events in patients with metastatic disease or skeletal-related cancers. Emerging data suggest that zoledronic acid may also exhibit anticancer properties. Zoledronic acid ± anticancer therapies in animal models inhibits soft-tissue tumor growth, decreases tumor cell proliferation, increases apoptosis, inhibits angiogenesis, alters tumor-associated macrophage function, and enhances immune surveillance. Data from in vitro and pilot studies suggest that zoledronic acid inhibits tumor cell dissemination in bone marrow, and early clinical data show that it may improve disease-related outcomes. Ongoing studies will further elucidate the role of zoledronic acid in cancer patients.
Collapse
|
35
|
Majumdar A, Sun Y, Shah M, Freel Meyers CL. Versatile (1)H-(31)P-(31)P COSY 2D NMR techniques for the characterization of polyphosphorylated small molecules. J Org Chem 2010; 75:3214-23. [PMID: 20408590 DOI: 10.1021/jo100042m] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Di- and triphosphorylated small molecules represent key intermediates in a wide range of biological and chemical processes. The importance of polyphosphorylated species in biology and medicine underscores the need to develop methods for the detection and characterization of this compound class. We have reported two-dimensional HPP-COSY spectroscopy techniques to identify diphosphate-containing metabolic intermediates at submillimolar concentrations in the methylerythritol phosphate (MEP) isoprenoid biosynthetic pathway. (1) In this work, we explore the scope of HPP-COSY-based techniques to characterize a diverse group of small organic molecules bearing di- and triphosphorylated moieties. These include molecules containing P-O-P and P-C-P connectivities, multivalent P(III)-O-P(V) phosphorus nuclei with widely separated chemical shifts, as well as virtually overlapping (31)P resonances exhibiting strong coupling effects. We also demonstrate the utility of these experiments to rapidly distinguish between mono- and diphosphates. A detailed protocol for optimizing these experiments to achieve best performance is presented.
Collapse
Affiliation(s)
- Ananya Majumdar
- The Johns Hopkins University Biomolecular NMR Center, JohnsHopkins School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | |
Collapse
|
36
|
[Antitumor properties of the bisphosphonate zoledronate and potential therapeutic implications in the clinic]. Bull Cancer 2010; 97:937-49. [PMID: 20595092 DOI: 10.1684/bdc.2010.1108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Zoledronate, just as other bisphosphonates, inhibit osteoclast mediated bone resorption. This is the reason why they are used in the treatment of bone metastasis, in order to block osteolysis. Zoledronate and some other bisphosphonates (clodronate, pamidronate, ibandronate, alendronate, risédronate, minodronate) also exhibit antitumor properties in vitro. They act directly on tumor cells by blocking tumor cell adhesion, invasion and proliferation, and by inducing tumor cell apoptosis. However, their high bone mineral affinity decreases their bioavailability to a significant extent and, thus, should weaken their in vivo antitumor potential. Despite of this, several studies (most of them being performed with zoledronate) show that bisphosphonates have an in vivo antitumor activity. This review focuses on zoledronate and on results obtained in several experimental models showing that this bisphosphonate interferes with the growth of tumors and metastases which are thriving in tissues others than the skeletal tissue. The significance of these findings is discussed in the light of several ongoing clinical trials which examine the benefits of using zoledronate and other bisphosphonates in the adjuvant treatment of cancers at an early stage of the disease.
Collapse
|
37
|
Multicenter, randomized, phase 2 study of zoledronic acid in combination with docetaxel and carboplatin in patients with unresectable stage IIIB or stage IV non-small cell lung cancer. Lung Cancer 2010; 67:330-8. [DOI: 10.1016/j.lungcan.2009.04.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Revised: 03/31/2009] [Accepted: 04/26/2009] [Indexed: 11/18/2022]
|
38
|
Langer C, Hirsh V. Skeletal morbidity in lung cancer patients with bone metastases: demonstrating the need for early diagnosis and treatment with bisphosphonates. Lung Cancer 2010; 67:4-11. [PMID: 19939491 DOI: 10.1016/j.lungcan.2009.08.020] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Revised: 08/21/2009] [Accepted: 08/27/2009] [Indexed: 11/18/2022]
Abstract
BACKGROUND The skeleton is one of the most frequent sites for metastases from non-small cell lung cancer (NSCLC), and skeletal-related events (SREs) can decrease quality of life (QOL). However, limited guidance exists regarding the diagnosis and treatment of bone metastases in patients with NSCLC. METHODS Data on the burden of skeletal morbidity and the diagnosis and treatment of bone metastases in patients with NSCLC were obtained from reviewing the published literature (PubMed) and presentations and abstracts from recent oncology congresses. RESULTS Bone metastases are common but underdiagnosed in patients with NSCLC. Most NSCLC patients with bone metastases develop > or =1 SRE during their lifetimes. As survival improves with new treatment modalities, the prevalence of SREs is likely to increase. Direct costs of SREs and their subsequent supportive care are approximately $28,000 per patient. Although bone metastases often are not diagnosed until after the onset of symptoms, early treatment can delay the onset of potentially debilitating SREs. In patients with NSCLC and other solid tumors (n=773), zoledronic acid (ZOL; 4 mg via 15-min infusion every 3 weeks) delayed the median time to first on-study SRE by >80 days compared with placebo (p=0.009). Moreover, ZOL significantly reduced the ongoing risk of SREs by 32% versus placebo (p=0.016). CONCLUSIONS Skeletal morbidity is an important concern in patients with NSCLC. The incidence of SREs is expected to increase as survival improves in this setting. Prevention of SREs with therapies such as ZOL may preserve patients' QOL and possibly reduce healthcare costs.
Collapse
Affiliation(s)
- Corey Langer
- Hematology-Oncology Division, University of Pennsylvania, 3400 Civic Center Blvd., 2 Perelman Center for Advanced Medicine, Philadelphia, PA 19104, USA.
| | | |
Collapse
|
39
|
Ottewell PD, Lefley DV, Cross SS, Evans CA, Coleman RE, Holen I. Sustained inhibition of tumor growth and prolonged survival following sequential administration of doxorubicin and zoledronic acid in a breast cancer model. Int J Cancer 2010; 126:522-32. [DOI: 10.1002/ijc.24756] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
40
|
Mitrofan LM, Pelkonen J, Mönkkönen J. The level of ATP analog and isopentenyl pyrophosphate correlates with zoledronic acid-induced apoptosis in cancer cells in vitro. Bone 2009; 45:1153-60. [PMID: 19699819 DOI: 10.1016/j.bone.2009.08.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Revised: 08/05/2009] [Accepted: 08/08/2009] [Indexed: 01/03/2023]
Abstract
Bisphosphonates are potent inhibitors of osteoclast function widely used to treat excessive bone resorption associated, e.g., with bone metastases. They have also antitumor activity. However, it is unclear whether this reflects an indirect effect via inhibition of bone resorption or a direct antitumor effect. Nitrogen-containing bisphosphonates (N-BPs), including zoledronic acid (ZOL), act by inhibiting farnesyl pyrophosphate synthase (FPPS). The mevalonate pathway is blocked and the accumulation of isopentenyl pyrophosphate (IPP) consequently occurs. IPP is conjugated to AMP to form a novel ATP analog (ApppI). The present study was undertaken to clarify whether IPP and/or ApppI has a direct involvement in apoptosis caused by ZOL in different cancer cell lines. There are marked differences in ZOL-induced ApppI formation between different cancer cell lines. On this basis, we selected three cancer cell lines that differ significantly from each other in their ZOL-induced IPP and ApppI accumulation: human estrogen-dependent (MCF7) and estrogen-independent (MDA-MB 436) breast cancer cell lines and a human myeloma cell line (RPMI 8226). The amount of IPP/ApppI correlated with the capacity of cells to undergo apoptosis. Geranylgeraniol (GGOH), an intermediate of mevalonate metabolism, blocks both IPP and ApppI formation and to some degree ZOL-induced apoptosis in a cell line-dependent manner. In addition, lovastatin (LOV), an inhibitor of the enzyme HMGCoA reductase, completely blocks IPP/ApppI formation as determined by mass spectrometry analysis, but enhances apoptosis. In conclusion, the current data suggest that ZOL-induced IPP/ApppI formation can contribute to ZOL-induced apoptosis. This mechanism and the inhibition of protein prenylation, both outcomes of FPPS inhibition in mevalonate pathway, seem to act in concert in ZOL-induced apoptosis in cancer cells.
Collapse
Affiliation(s)
- Laura M Mitrofan
- Faculty of Pharmacy, Department of Pharmaceutics, University of Kuopio, Finland
| | | | | |
Collapse
|
41
|
Bisphosphonate Use in Patients with Lung Cancer and Bone Metastases: Recommendations of a European Expert Panel. J Thorac Oncol 2009; 4:1280-8. [DOI: 10.1097/jto.0b013e3181b68e5a] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
42
|
Rachner TD, Singh SK, Schoppet M, Benad P, Bornhäuser M, Ellenrieder V, Ebert R, Jakob F, Hofbauer LC. Zoledronic acid induces apoptosis and changes the TRAIL/OPG ratio in breast cancer cells. Cancer Lett 2009; 287:109-16. [PMID: 19577359 DOI: 10.1016/j.canlet.2009.06.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2009] [Revised: 05/29/2009] [Accepted: 06/04/2009] [Indexed: 01/06/2023]
Abstract
Breast cancer has a propensity to metastasize to bone, thus causing pathological fractures. Bisphosphonates are established drugs in the treatment of bone metastasis that inhibit osteoclast activity and interrupt the vicious cycle of osteoclast-tumor cell interactions. We evaluated the direct effects of zoledronic acid on estrogen receptor (ER)-negative MDA-MB-231 and ER-positive MCF-7 breast cancer cells. While zoledronic acid (100 microM) inhibited MDA-MB-231 cell proliferation after 72 h, and induced apoptosis via activation of caspase-3 and -7, it had only minor effects on MCF-7 cells. In addition, zoledronic acid induced apoptosis by up-regulating TNF-related apoptosis-inducing ligand (TRAIL) in MDA-MB-231 cells (p<0.01), but had no effect on the expression of its decoy receptor osteoprotegerin (OPG). In MCF-7 cells, both cytokines were suppressed by zoledronic acid. In conclusion, zoledronic acid enhanced the TRAIL-to-OPG ratio in TRAIL-sensitive MDA-MB-231 cells, indicating that the TRAIL/OPG cytokine system is a bisphosphonate-responsive target in breast cancer.
Collapse
Affiliation(s)
- Tilman D Rachner
- Division of Gastroenterology and Endocrinology, Department of Medicine, Philipps-University, Marburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Chang JWC, Hsieh JJ, Shen YC, Yeh KY, Wang CH, Li YY, Hsu T. Bisphosphonate zoledronic acid enhances the inhibitory effects of gefitinib on EGFR-mutated non-small cell lung carcinoma cells. Cancer Lett 2009; 278:17-26. [DOI: 10.1016/j.canlet.2008.12.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Revised: 11/21/2008] [Accepted: 12/10/2008] [Indexed: 01/14/2023]
|
44
|
Romani AA, Desenzani S, Morganti MM, La Monica S, Borghetti AF, Soliani P. Zoledronic acid determines S-phase arrest but fails to induce apoptosis in cholangiocarcinoma cells. Biochem Pharmacol 2009; 78:133-41. [PMID: 19464430 DOI: 10.1016/j.bcp.2009.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Revised: 03/13/2009] [Accepted: 04/02/2009] [Indexed: 02/07/2023]
Abstract
Cholangiocarcinoma is the second most common primary hepatic neoplasia and the only curative therapy is surgical resection or liver transplantation. Biphosphonates (BPs) are an emerging class of drugs widely used to treat bone diseases and also appear to possess direct antitumor activity. In two human cholangiocarcinoma cell lines (TFK-1 and EGI-1) we investigated, for the first time, the activity of zoledronic acid by determining proliferation, cell cycle analysis and apoptosis. The results obtained indicate that zoledronic acid induces cell-narrowing and growth inhibition, both reversed by 25 microM GGOH, and significantly affects the colony-forming ability of these cells. The inhibition by zoledronic acid of Rap1A prenylation was reversed in cell co-treated with GGOH. At 10-50 microM zoledronic acid exerted an S-phase cell cycle arrest which was confirmed by changes in the level of cyclins and of regulators p27(KIP1) and pRb. Interestingly, the expression level of cyclin A (putative S-phase marker) shows a dose-dependent increment in contrast to the decrement of cyclin D1 (putative G1 phase marker). However, neither hypodiploid cells nor cleaved PARP or caspase-3 was detected. The lack of TP53 or loss of its function, the large constitutive expressions of anti-apoptotic proteins Bcl-xL and HSP27 together with the low level of the pro-apoptotic Bax are the likely factors which protect cells from apoptosis. In conclusion, our study indicates that zoledronic acid induces S-phase arrest and cell-narrowing, both reversed by GGOH and, by changing the delicate balance between pro- and anti-apoptotic proteins, allows survival of cholangiocarcinoma cells.
Collapse
Affiliation(s)
- Antonello A Romani
- Dipartimento di Medicina Sperimentale, Sezione di Patologia Molecolare ed Immunologia, Università degli Studi di Parma, via Volturno 39, 43100 Parma, Italy
| | | | | | | | | | | |
Collapse
|
45
|
Zoledronate inhibits the proliferation, adhesion and migration of vascular smooth muscle cells. Eur J Pharmacol 2008; 602:124-31. [PMID: 19000670 DOI: 10.1016/j.ejphar.2008.10.043] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2008] [Revised: 09/23/2008] [Accepted: 10/15/2008] [Indexed: 11/20/2022]
Abstract
Bisphosphonates, which are extensively used in bone-related disorders, have been reported to inhibit atherosclerosis and neointimal hyperplasia. In the present study, we investigated the effects of a bisphosphonate, zoledronate, on the proliferation, adhesion, migration and microstructure of vascular smooth muscle cells (VSMCs) from Sprague-Dawley rats. It was shown that zoledronate suppressed VSMCs proliferation after 48 h cultivation in a dose depend manner, most obviously at concentrations above 10 microM. Cell cycle analysis indicated that zoledronate inhibited the proliferation of VSMCs via cell cycle arrest at S/G2/M phase. This inhibition was not associated with cell death. In a modified Boyden chamber model, it was shown that zoledronate dose-dependently inhibited VSMCs adhesion to collagen and migration stimulated by platelet-derived growth factor-BB. Western blot analysis suggested that zoledronate significantly inhibited the phosphorylation of focal adhesion kinase. Furthermore, we observed that more and more VSMCs changed from a bipolar appearance to a globular shape under inverted light microscope as zoledronate concentration increased from 0.1 to 100 microM. Images under transmission electron microscope confirmed this morphological change, and many electron density bodies were observed in zoledronate-treated VSMCs. These findings indicated that bisphosphonates' effects of suppressing atherosclerosis and neointimal hyperplasia might be due to inhibition of VSMCs, at least for zoledronate.
Collapse
|
46
|
Ottewell PD, Mönkkönen H, Jones M, Lefley DV, Coleman RE, Holen I. Antitumor effects of doxorubicin followed by zoledronic acid in a mouse model of breast cancer. J Natl Cancer Inst 2008; 100:1167-78. [PMID: 18695136 DOI: 10.1093/jnci/djn240] [Citation(s) in RCA: 181] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The potent antiresorptive drug zoledronic acid (Zol) enhances the antitumor effects of chemotherapy agents in vitro. We investigated the effects of clinically achievable doses of doxorubicin (Dox) and Zol, given alone, in sequence, and in combination, on the growth of established breast tumors in vivo. METHODS Female MF1 nude mice were inoculated subcutaneously with 5 x 10(5) human breast cancer MDA-MB-436 cells that stably expressed green fluorescent protein (ie, MDA-G8 cells). Beginning on day 7 after tumor cell injection, the mice were injected weekly for 6 weeks with saline, Dox (2 mg/kg body weight via intravenous injection), Zol (100 microg/kg body weight via intraperitoneal injection), Dox plus Zol, Zol followed 24 hours later by Dox, or Dox followed 24 hours later by Zol (n = 8-9 mice per group). The effects of treatment on tumor growth were determined by measuring tumor volume; on tumor cell apoptosis and proliferation by immunohistochemistry using antibodies for caspase-3 and Ki-67, respectively; and on bone by microcomputed tomography and bone histomorphometry. All P values are two-sided. RESULTS Treatment with Dox or Zol alone or Zol followed 24 hours later by Dox did not statistically significantly decrease final tumor volume compared with saline. Mice treated with Dox plus Zol had statistically significantly smaller final tumor volumes than those treated with Dox alone (mean = 122 mm(3) vs 328 mm(3), difference = 206 mm(3), 95% confidence interval [CI] = 78 to 335 mm(3), P < .001), with Zol alone (122 mm(3) vs 447 mm(3), difference = 325 mm(3), 95% CI = 197 to 454 mm(3), P < .001), or with Zol followed 24 hours later by Dox (122 mm(3) vs 418 mm(3), difference = 296 mm(3), 95% CI = 168 to 426 mm(3), P < .001). Treatment with Dox followed 24 hours later by Zol almost completely abolished tumor growth. Tumors from mice that were treated with Dox followed by Zol had more caspase-3-positive cells than tumors from mice treated with saline (mean number of caspase-3-positive cells per square millimeter: 605.0 vs 82.19, difference = 522.8, 95% CI = 488.2 to 557.4, P < .001), with Zol alone (605.0 vs 98.44, difference = 506.6, 95% CI = 472.0 to 541.2, P < .001), or with Zol followed by Dox (605.0 vs 103.1, difference = 501.9, 95% CI = 467.3 to 536.5, P < .001). The treatment-induced increase in the number of caspase-3-positive cells was mirrored by a decrease in the number of tumor cells positive for the proliferation marker Ki-67. No evidence of bone disease was detected in any of the treatment groups following microcomputed tomography and histological analysis of bone. CONCLUSION Sequential treatment with Dox followed by Zol elicited substantial antitumor effects in subcutaneous breast tumors in vivo, in the absence of bone disease.
Collapse
Affiliation(s)
- Penelope D Ottewell
- Academic Unit of Clinical Oncology, School of Medicine and Biomedical Sciences, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
| | | | | | | | | | | |
Collapse
|
47
|
The bisphosphonate zoledronic acid inhibits the growth of HCT-116 colon carcinoma cells and induces tumor cell apoptosis. Apoptosis 2008; 13:782-9. [PMID: 18437576 DOI: 10.1007/s10495-008-0211-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Besides its preventive action on bone resorption the third generation bisphosphonate zoledronic acid (ZOL) has been shown to display potent inhibitory action on the formation of bone metastases of various human cancers. Recent research also indicates an antitumoral effect on primary tumors and visceral metastases. Here we investigate for the first time the effect of ZOL on the human colon carcinoma cell line HCT-116. ZOL strongly inhibited the proliferation and soft agar colony formation of HCT-116 cells and caused a G1 cell cycle arrest in a population of ZOL treated cells. This cell cycle arrest was accompanied by an induction of apoptosis via a caspase dependent mechanism. Activation of Caspases 3, 7, 8 and 9, cleavage of PARP as well as the release of cytochrome C into the cytosol were detected in HCT-116 cells treated with low micromolar concentrations of ZOL. The induction of the mitochondrial pathway of apoptosis was accompanied by a translocation of Bax into the mitochondria, Bid activation and a decrease of overall Bcl-2 expression. We also detected a cytosolic increase of apoptosis inducing factor (AIF), a trigger of caspase-independent apoptosis. Taken together, our data indicate a potent antitumoral and apoptosis inducing effect of ZOL on HCT-116 colon carcinoma cells.
Collapse
|